Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Pharm Res ; 40(10): 2355-2370, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37131104

ABSTRACT

BACKGROUND: Spray-drying is considered a promising alternative drying method to lyophilization (freeze-drying) for therapeutic proteins. Particle counts in reconstituted solutions of dried solid dosage forms of biologic drug products are closely monitored to ensure product quality. We found that high levels of particles formed after reconstitution of protein powders that had been spray-dried under suboptimal conditions. METHODS: Visible and subvisible particles were evaluated. Soluble proteins in solution before spray-drying and in the reconstituted solution of spray-dried powder were analyzed for their monomer content levels and melting temperatures. Insoluble particles were collected and analyzed by Fourier transform infrared microscopy (FTIR), and further analyzed with hydrogen-deuterium exchange (HDX). RESULTS: Particles observed after reconstitution were shown not to be undissolved excipients. FTIR confirmed their identity as proteinaceous in nature. These particles were therefore considered to be insoluble protein aggregates, and HDX was applied to investigate the mechanism underlying aggregate formation. Heavy-chain complementarity-determining region 1 (CDR-1) in the aggregates showed significant protection by HDX, suggesting CDR-1 was critical for aggregate formation. In contrast, various regions became more conformationally dynamic globally, suggesting the aggregates have lost protein structural integrity and partially unfolded after spray-drying. DISCUSSION: The spray-drying process could have disrupted the higher-order structure of proteins and exposed the hydrophobic residues in CDR-1 of the heavy chain, contributing to the formation of aggregate through hydrophobic interactions upon reconstitution of spray-dried powder. These results can contribute to efforts to design spray-dry resilient protein constructs and improve the robustness of the spray-drying process.


Subject(s)
Microscopy , Proteins , Powders/chemistry , Freeze Drying , Particle Size
2.
J Proteome Res ; 20(6): 3150-3164, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34008986

ABSTRACT

Citrullination is an important post-translational modification implicated in many diseases including rheumatoid arthritis (RA), Alzheimer's disease, and cancer. Neutrophil and mast cells have different expression profiles for protein-arginine deiminases (PADs), and ionomycin-induced activation makes them an ideal cellular model to study proteins susceptible to citrullination. We performed high-resolution mass spectrometry and stringent data filtration to identify citrullination sites in neutrophil and mast cells treated with and without ionomycin. We identified a total of 833 validated citrullination sites on 395 proteins. Several of these citrullinated proteins are important components of pathways involved in innate immune responses. Using this benchmark primary sequence data set, we developed machine learning models to predict citrullination in neutrophil and mast cell proteins. We show that our models predict citrullination likelihood with 0.735 and 0.766 AUCs (area under the receiver operating characteristic curves), respectively, on independent validation sets. In summary, this study provides the largest number of validated citrullination sites in neutrophil and mast cell proteins. The use of our novel motif analysis approach to predict citrullination sites will facilitate the discovery of novel protein substrates of protein-arginine deiminases (PADs), which may be key to understanding immunopathologies of various diseases.


Subject(s)
Citrullination , Mast Cells , Citrulline/metabolism , Ionomycin/pharmacology , Machine Learning , Mass Spectrometry , Mast Cells/metabolism , Neutrophils/metabolism , Protein-Arginine Deiminases/genetics
3.
Proc Natl Acad Sci U S A ; 114(25): 6557-6562, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28584102

ABSTRACT

Strains of the Burkholderia cepacia complex (Bcc) are Gram-negative opportunisitic bacteria that are capable of causing serious diseases, mainly in immunocompromised individuals. Bcc pathogens are intrinsically resistant to multiple antibiotics, including ß-lactams, aminoglycosides, fluoroquinolones, and polymyxins. They are major pathogens in patients with cystic fibrosis (CF) and can cause severe necrotizing pneumonia, which is often fatal. Hopanoid biosynthesis is one of the major mechanisms involved in multiple antimicrobial resistance of Bcc pathogens. The hpnN gene of B. multivorans encodes an integral membrane protein of the HpnN family of transporters, which is responsible for shuttling hopanoids to the outer membrane. Here, we report crystal structures of B. multivorans HpnN, revealing a dimeric molecule with an overall butterfly shape. Each subunit of the transporter contains 12 transmembrane helices and two periplasmic loops that suggest a plausible pathway for substrate transport. Further analyses indicate that HpnN is capable of shuttling hopanoid virulence factors from the outer leaflet of the inner membrane to the periplasm. Taken together, our data suggest that the HpnN transporter is critical for multidrug resistance and cell wall remodeling in Burkholderia.


Subject(s)
Burkholderia cepacia complex/chemistry , Membrane Transport Proteins/chemistry , Crystallography, X-Ray/methods , Periplasm/chemistry , Virulence Factors/chemistry
4.
J Biol Chem ; 290(47): 28559-28574, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26396194

ABSTRACT

The mycobacterial cell wall is critical to the virulence of these pathogens. Recent work shows that the MmpL (mycobacterial membrane protein large) family of transporters contributes to cell wall biosynthesis by exporting fatty acids and lipidic elements of the cell wall. The expression of the Mycobacterium tuberculosis MmpL proteins is controlled by a complex regulatory network, including the TetR family transcriptional regulators Rv3249c and Rv1816. Here we report the crystal structures of these two regulators, revealing dimeric, two-domain molecules with architecture consistent with the TetR family of regulators. Buried extensively within the C-terminal regulatory domains of Rv3249c and Rv1816, we found fortuitous bound ligands, which were identified as palmitic acid (a fatty acid) and isopropyl laurate (a fatty acid ester), respectively. Our results suggest that fatty acids may be the natural ligands of these regulatory proteins. Using fluorescence polarization and electrophoretic mobility shift assays, we demonstrate the recognition of promoter and intragenic regions of multiple mmpL genes by these proteins. Binding of palmitic acid renders these regulators incapable of interacting with their respective operator DNAs, which will result in derepression of the corresponding mmpL genes. Taken together, these experiments provide new perspectives on the regulation of the MmpL family of transporters.


Subject(s)
Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/chemistry , Crystallography, X-Ray , Membrane Transport Proteins/chemistry , Protein Conformation
5.
Biometals ; 26(4): 593-607, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23657864

ABSTRACT

Resistance-nodulation-cell division (RND) superfamily efflux systems are responsible for the active transport of toxic compounds from the Gram-negative bacterial cell. These pumps typically assemble as tripartite complexes, spanning the inner and outer membranes of the cell envelope. In Escherichia coli, the CusC(F)BA complex, which exports copper(I) and silver(I) and mediates resistance to these two metal ions, is the only known RND transporter with a specificity for heavy metals. We have determined the crystal structures of both the inner membrane pump CusA and membrane fusion protein CusB, as well as the adaptor-transporter CusBA complex formed by these two efflux proteins. In addition, the crystal structures of the outer membrane channel CusC and the periplasmic metallochaperone CusF have been resolved. Based on these structures, the entire assembled model of the tripartite efflux system has been developed, and this efflux complex should be in the form of CusC3-CusB6-CusA3. It has been shown that CusA utilizes methionine clusters to bind and export Cu(I) and Ag(I). This pump is likely to undergo a conformational change, and utilize a relay network of methionine clusters as well as conserved charged residues to extrude the metal ions from the bacterial cell.


Subject(s)
Metals, Heavy/metabolism , Biological Transport , Copper/metabolism , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Models, Theoretical , Silver/metabolism
6.
J Pharm Sci ; 111(11): 3009-3016, 2022 11.
Article in English | MEDLINE | ID: mdl-35940243

ABSTRACT

During early stage development of a therapeutic IgG1 monoclonal antibody, high levels of low molecular weight (LMW) peaks were observed by high performance size-exclusion chromatography and capillary electrophoresis. Further characterization of the LMW peak enriched HPSEC fractions using reversed phase liquid chromatography coupled to mass spectrometry showed these LMW species were 47 kDa and 50 kDa in size. However, the measured masses could not be matched to any fragments resulting from peptide bond hydrolysis. To identify these unknown LMW species, molecular characterization methods were employed, including high-throughput sequencing of RNA. Transcriptomic analysis revealed the LMW species were generated by mis-splicing events in the heavy chain transcript, which produced truncated heavy chain products that assembled with the light chain to mimic the appearance of fragments identified by routine purity assays. In an effort to improve product quality, an optimized purification process was developed. Characterization of the process intermediates confirmed removal of both LMW species by the optimized process. Our study demonstrates that deep-dive analytical characterization of biotherapeutics is critical to ensure product quality and inform process development. Transcriptomic analysis tools can help identify the cause of unknown species, and plays a key role in product and process characterization.


Subject(s)
Antibodies, Monoclonal , Chromatography, Reverse-Phase , Antibodies, Monoclonal/chemistry , Chromatography, Reverse-Phase/methods , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Peptides , RNA
7.
Mol Ther Methods Clin Dev ; 21: 466-477, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-33898635

ABSTRACT

Photooxidation of methionine (Met) and tryptophan (Trp) residues is common and includes major degradation pathways that often pose a serious threat to the success of therapeutic proteins. Oxidation impacts all steps of protein production, manufacturing, and shelf life. Prediction of oxidation liability as early as possible in development is important because many more candidate drugs are discovered than can be tested experimentally. Undetected oxidation liabilities necessitate expensive and time-consuming remediation strategies in development and may lead to good drugs reaching patients slowly. Conversely, sites mischaracterized as oxidation liabilities could result in overengineering and lead to good drugs never reaching patients. To our knowledge, no predictive model for photooxidation of Met or Trp is currently available. We applied the random forest machine learning algorithm to in-house liquid chromatography-tandem mass spectrometry (LC-MS/MS) datasets (Met, n = 421; Trp, n = 342) of tryptic therapeutic protein peptides to create computational models for Met and Trp photooxidation. We show that our machine learning models predict Met and Trp photooxidation likelihood with 0.926 and 0.860 area under the curve (AUC), respectively, and Met photooxidation rate with a correlation coefficient (Q2) of 0.511 and root-mean-square error (RMSE) of 10.9%. We further identify important physical, chemical, and formulation parameters that influence photooxidation. Improvement of biopharmaceutical liability predictions will result in better, more stable drugs, increasing development throughput, product quality, and likelihood of clinical success.

8.
Mol Ther Methods Clin Dev ; 19: 374, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33145373

ABSTRACT

[This corrects the article DOI: 10.1016/j.omtm.2019.09.008.].

9.
Mol Ther Methods Clin Dev ; 15: 264-274, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31890727

ABSTRACT

The spontaneous conversion of asparagine residues to aspartic acid or iso-aspartic acid, via deamidation, is a major pathway of protein degradation and is often seriously disruptive to biological systems. Deamidation has been shown to negatively affect both in vitro stability and in vivo biological function of diverse classes of proteins. During protein therapeutics development, deamidation liabilities that are overlooked necessitate expensive and time-consuming remediation strategies, sometimes leading to termination of the project. In this paper, we apply machine learning to a large (n = 776) liquid chromatography-tandem mass spectrometry (LC-MS/MS) dataset of monoclonal antibody peptides to create computational models for the post-translational modification asparagine deamidation, using the random decision forest method. We show that our categorical model predicts antibody deamidation with nearly 5% increased accuracy and 0.2 MCC over the best currently available models. Surprisingly, our model also paces or outperforms advanced and conventional models on an independent non-antibody dataset. In addition to deamidation probability, we are able to accurately predict deamidation rate (R2 = 0.963 and Q2 = 0.822), a capability with no peer in current models. This method should enable significant improvement in protein candidate selection, especially in biopharmaceutical development, and can be applied with similar accuracy to enzymes, monoclonal antibodies, next-generation formats, vaccine component antigens, and gene therapy vectors such as adeno-associated virus.

10.
Methods Mol Biol ; 1700: 59-70, 2018.
Article in English | MEDLINE | ID: mdl-29177825

ABSTRACT

Crystallization is one of the most successful techniques used to determine protein structure, especially for membrane proteins. However, the application of this technique is not straightforward and often hampered by the difficulties associated with expression, purification, and crystallization. Here we present our protocol and methodology for crystallizing the CusBA adaptor-transporter complex of Escherichia coli. Using these procedures, we were able to produce the first co-crystal structure of a resistance-nodulation-cell division (RND) transporter in complex with its associated membrane fusion protein.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Membrane Transport Proteins/chemistry , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Metals, Heavy/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Protein Binding , Protein Conformation
11.
Nat Commun ; 8(1): 171, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28761097

ABSTRACT

Resistance-nodulation-cell division efflux pumps are integral membrane proteins that catalyze the export of substrates across cell membranes. Within the hydrophobe-amphiphile efflux subfamily, these resistance-nodulation-cell division proteins largely form trimeric efflux pumps. The drug efflux process has been proposed to entail a synchronized motion between subunits of the trimer to advance the transport cycle, leading to the extrusion of drug molecules. Here we use X-ray crystallography and single-molecule fluorescence resonance energy transfer imaging to elucidate the structures and functional dynamics of the Campylobacter jejuni CmeB multidrug efflux pump. We find that the CmeB trimer displays a very unique conformation. A direct observation of transport dynamics in individual CmeB trimers embedded in membrane vesicles indicates that each CmeB subunit undergoes conformational transitions uncoordinated and independent of each other. On the basis of our findings and analyses, we propose a model for transport mechanism where CmeB protomers function independently within the trimer.Multidrug efflux pumps significantly contribute for bacteria resistance to antibiotics. Here the authors present the structure of Campylobacter jejuni CmeB pump combined with functional FRET assays to propose a transport mechanism where each CmeB protomers is functionally independent from the trimer.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Bacterial Proteins/genetics , Campylobacter jejuni/genetics , Crystallography, X-Ray , Drug Resistance, Multiple, Bacterial/genetics , Fluorescence Resonance Energy Transfer , Membrane Transport Proteins/genetics , Protein Conformation , Protein Structure, Secondary
12.
Protein Sci ; 25(2): 322-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26443496

ABSTRACT

The AbgT family of transporters was thought to contribute to bacterial folate biosynthesis by importing the catabolite p-aminobenzoyl-glutamate for producing this essential vitamin. Approximately 13,000 putative transporters of the family have been identified. However, before our work, no structural information was available and even functional data were minimal for this family of membrane proteins. To elucidate the structure and function of the AbgT family of transporters, we recently determined the X-ray structures of the full-length Alcanivorax borkumensis YdaH and Neisseria gonorrhoeae MtrF membrane proteins. The structures reveal that these two transporters assemble as dimers with architectures distinct from all other families of transporters. Both YdaH and MtrF are bowl-shaped dimers with a solvent-filled basin extending from the cytoplasm halfway across the membrane bilayer. The protomers of YdaH and MtrF contain nine transmembrane helices and two hairpins. These structures directly suggest a plausible pathway for substrate transport. A combination of the crystal structure, genetic analysis and substrate accumulation assay indicates that both YdaH and MtrF behave as exporters, capable of removing the folate metabolite p-aminobenzoic acid from bacterial cells. Further experimental data based on drug susceptibility and radioactive transport assay suggest that both YdaH and MtrF participate as antibiotic efflux pumps, importantly mediating bacterial resistance to sulfonamide antimetabolite drugs. It is possible that many of these AbgT-family transporters act as exporters, thereby conferring bacterial resistance to sulfonamides. The AbgT-family transporters may be important targets for the rational design of novel antibiotics to combat bacterial infections.


Subject(s)
Antimetabolites/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Bacteria/chemistry , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biological Transport , Crystallography, X-Ray , Folic Acid/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
13.
Protein Sci ; 24(11): 1720-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26258953

ABSTRACT

It is widely accepted that the increased use of antibiotics has resulted in bacteria with developed resistance to such treatments. These organisms are capable of forming multi-protein structures that bridge both the inner and outer membrane to expel diverse toxic compounds directly from the cell. Proteins of the resistance nodulation cell division (RND) superfamily typically assemble as tripartite efflux pumps, composed of an inner membrane transporter, a periplasmic membrane fusion protein, and an outer membrane factor channel protein. These machines are the most powerful antimicrobial efflux machinery available to bacteria. In Escherichia coli, the CusCFBA complex is the only known RND transporter with a specificity for heavy metals, detoxifying both Cu(+) and Ag(+) ions. In this review, we discuss the known structural information for the CusCFBA proteins, with an emphasis on their assembly, interaction, and the relationship between structure and function.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Metals, Heavy/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Metals, Heavy/chemistry , Models, Molecular
14.
Methods Enzymol ; 557: 363-92, 2015.
Article in English | MEDLINE | ID: mdl-25950974

ABSTRACT

X-ray crystallography remains the most robust method to determine protein structure at the atomic level. However, the bottlenecks of protein expression and purification often discourage further study. In this chapter, we address the most common problems encountered at these stages. Based on our experiences in expressing and purifying antimicrobial efflux proteins, we explain how a pure and homogenous protein sample can be successfully crystallized by the vapor diffusion method. We present our current protocols and methodologies for this technique. Case studies show step-by-step how we have overcome problems related to expression and diffraction, eventually producing high-quality membrane protein crystals for structural determinations. It is our hope that a rational approach can be made of the often anecdotal process of membrane protein crystallization.


Subject(s)
Crystallography, X-Ray/methods , Membrane Proteins/chemistry , Animals , Bacteria/chemistry , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Detergents/chemistry , Diffusion , Gene Expression , Humans , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Models, Molecular , Volatilization
15.
Nat Commun ; 6: 6874, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25892120

ABSTRACT

The potential of the folic acid biosynthesis pathway as a target for the development of antibiotics has been clinically validated. However, many pathogens have developed resistance to these antibiotics, prompting a re-evaluation of potential drug targets within the pathway. The ydaH gene of Alcanivorax borkumensis encodes an integral membrane protein of the AbgT family of transporters for which no structural information was available. Here we report the crystal structure of A. borkumensis YdaH, revealing a dimeric molecule with an architecture distinct from other families of transporters. YdaH is a bowl-shaped dimer with a solvent-filled basin extending from the cytoplasm to halfway across the membrane bilayer. Each subunit of the transporter contains nine transmembrane helices and two hairpins that suggest a plausible pathway for substrate transport. Further analyses also suggest that YdaH could act as an antibiotic efflux pump and mediate bacterial resistance to sulfonamide antimetabolite drugs.


Subject(s)
Alcanivoraceae/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Alcanivoraceae/drug effects , Alcanivoraceae/genetics , Anti-Infective Agents/metabolism , Bacterial Proteins/genetics , Carrier Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Folic Acid/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Sulfamethazine/metabolism
16.
Cell Rep ; 11(1): 61-70, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25818299

ABSTRACT

Neisseria gonorrhoeae is an obligate human pathogen and the causative agent of the sexually transmitted disease gonorrhea. The control of this disease has been compromised by the increasing proportion of infections due to antibiotic-resistant strains, which are growing at an alarming rate. N. gonorrhoeae MtrF is an integral membrane protein that belongs to the AbgT family of transporters for which no structural information is available. Here, we describe the crystal structure of MtrF, revealing a dimeric molecule with architecture distinct from all other families of transporters. MtrF is a bowl-shaped dimer with a solvent-filled basin extending from the cytoplasm to halfway across the membrane bilayer. Each subunit of the transporter contains nine transmembrane helices and two hairpins, posing a plausible pathway for substrate transport. A combination of the crystal structure and biochemical functional assays suggests that MtrF is an antibiotic efflux pump mediating bacterial resistance to sulfonamide antimetabolite drugs.


Subject(s)
Bacterial Proteins/chemistry , Drug Resistance, Bacterial/genetics , Gonorrhea/microbiology , Neisseria gonorrhoeae/chemistry , Repressor Proteins/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/metabolism , Crystallography, X-Ray , Gene Expression Regulation, Bacterial/drug effects , Gonorrhea/drug therapy , Gonorrhea/genetics , Humans , Models, Molecular , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/genetics , Protein Conformation , Repressor Proteins/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/therapeutic use
17.
Protein Sci ; 24(12): 1942-55, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26362239

ABSTRACT

Mycobacterium tuberculosis is a pathogenic bacterial species, which is neither Gram positive nor Gram negative. It has a unique cell wall, making it difficult to kill and conferring resistance to antibiotics that disrupt cell wall biosynthesis. Thus, the mycobacterial cell wall is critical to the virulence of these pathogens. Recent work shows that the mycobacterial membrane protein large (MmpL) family of transporters contributes to cell wall biosynthesis by exporting fatty acids and lipidic elements of the cell wall. The expression of the Mycobacterium tuberculosis MmpL proteins is controlled by a complicated regulatory network system. Here we report crystallographic structures of two forms of the TetR-family transcriptional regulator Rv0302, which participates in regulating the expression of MmpL proteins. The structures reveal a dimeric, two-domain molecule with architecture consistent with the TetR family of regulators. Comparison of the two Rv0302 crystal structures suggests that the conformational changes leading to derepression may be due to a rigid body rotational motion within the dimer interface of the regulator. Using fluorescence polarization and electrophoretic mobility shift assays, we demonstrate the recognition of promoter and intragenic regions of multiple mmpL genes by this protein. In addition, our isothermal titration calorimetry and electrophoretic mobility shift experiments indicate that fatty acids may be the natural ligand of this regulator. Taken together, these experiments provide new perspectives on the regulation of the MmpL family of transporters.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/genetics , Cell Wall/chemistry , Cell Wall/metabolism , Crystallography, X-Ray , Fatty Acids/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/metabolism , Models, Molecular , Mycobacterium tuberculosis/chemistry , Promoter Regions, Genetic , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary
18.
Annu Rev Biophys ; 43: 93-117, 2014.
Article in English | MEDLINE | ID: mdl-24702006

ABSTRACT

Infections caused by bacteria are a leading cause of death worldwide. Although antibiotics remain a key clinical therapy, their effectiveness has been severely compromised by the development of drug resistance in bacterial pathogens. Multidrug efflux transporters--a common and powerful resistance mechanism--are capable of extruding a number of structurally unrelated antimicrobials from the bacterial cell, including antibiotics and toxic heavy metal ions, facilitating their survival in noxious environments. Transporters of the resistance-nodulation-cell division (RND) superfamily typically assemble as tripartite efflux complexes spanning the inner and outer membranes of the cell envelope. In Escherichia coli, the CusCFBA complex, which mediates resistance to copper(I) and silver(I) ions, is the only known RND transporter specific to heavy metals. Here, we describe the current knowledge of individual pump components of the Cus system, a paradigm for efflux machinery, and speculate on how RND pumps assemble to fight diverse antimicrobials.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Membrane Transport Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry
19.
Protein Sci ; 23(7): 954-61, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24753291

ABSTRACT

As one of the world's most prevalent enteric pathogens, Campylobacter jejuni is a major causative agent of human enterocolitis and is responsible for more than 400 million cases of diarrhea each year. The impact of this pathogen on children is of particular significance. Campylobacter has developed resistance to many antimicrobial agents via multidrug efflux machinery. The CmeABC tripartite multidrug efflux pump, belonging to the resistance-nodulation-cell division (RND) superfamily, plays a major role in drug resistant phenotypes of C. jejuni. This efflux complex spans the entire cell envelop of C. jejuni and mediates resistance to various antibiotics and toxic compounds. We here report the crystal structure of C. jejuni CmeC, the outer membrane component of the CmeABC tripartite multidrug efflux system. The structure reveals a possible mechanism for substrate export.


Subject(s)
Bacterial Proteins/chemistry , Campylobacter jejuni/metabolism , Crystallography, X-Ray , Ion Channels/chemistry , Bacterial Proteins/genetics , Campylobacter jejuni/genetics , Cysteine/metabolism , Models, Molecular , Protein Conformation , Protein Structure, Secondary
20.
PLoS One ; 9(6): e97475, 2014.
Article in English | MEDLINE | ID: mdl-24901251

ABSTRACT

Active efflux of antimicrobial agents is one of the most important strategies used by bacteria to defend against antimicrobial factors present in their environment. Mediating many cases of antibiotic resistance are transmembrane efflux pumps, composed of one or more proteins. The Neisseria gonorrhoeae MtrCDE tripartite multidrug efflux pump, belonging to the hydrophobic and amphiphilic efflux resistance-nodulation-cell division (HAE-RND) family, spans both the inner and outer membranes of N. gonorrhoeae and confers resistance to a variety of antibiotics and toxic compounds. We here describe the crystal structure of N. gonorrhoeae MtrE, the outer membrane component of the MtrCDE tripartite multidrug efflux system. This trimeric MtrE channel forms a vertical tunnel extending down contiguously from the outer membrane surface to the periplasmic end, indicating that our structure of MtrE depicts an open conformational state of this channel.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Models, Molecular , Neisseria gonorrhoeae/metabolism , Protein Conformation , Bacterial Outer Membrane Proteins/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL